hosted by
publicationslist.org
    

Federica Di Nicolantonio

Dr Federica Di Nicolantonio, PharmD, PhD
University of Torino, Department of Oncology,
IRCC at Candiolo, Laboratory of Pharmacogenomics, SP 142 Km 3.95
10060 Candiolo - TURIN - Italy


federicadn@hotmail.com

Journal articles

2013
Federica Di Nicolantonio, Alberto Bardelli (2013)  Mouse Models of Kras-Mutant Colorectal Cancer: Valuable GEMMs for Drug Testing?   Clinical cancer research : an official journal of the American Association for Cancer Research 19: 11. 2794-2796 Jun  
Abstract: The development of effective therapies for colorectal cancer depends on the ability of preclinical models to faithfully recapitulate the molecular and biologic behavior of human tumors. This study reports on the characterization of colorectal genetically engineered mouse models and their derivative cell lines carrying wild-type or oncogenic Kras with concomitant Apc and p53 loss. Clin Cancer Res; 19(11); 2794-6. ©2013 AACR.
Notes:
Alberto Bardelli, Simona Corso, Andrea Bertotti, Sebastijan Hobor, Emanuele Valtorta, Giulia Siravegna, Andrea Sartore-Bianchi, Elisa Scala, Andrea Cassingena, Davide Zecchin, Maria Apicella, Giorgia Migliardi, Francesco Galimi, Calogero Lauricella, Carlo Zanon, Timothy Perera, Silvio Veronese, Giorgio Corti, Alessio Amatu, Marcello Gambacorta, Luis A Diaz, Mark Sausen, Victor E Velculescu, Paolo Comoglio, Livio Trusolino, Federica Di Nicolantonio, Silvia Giordano, Salvatore Siena (2013)  Amplification of the MET Receptor Drives Resistance to Anti-EGFR Therapies in Colorectal Cancer.   Cancer discovery 3: 6. 658-673 Jun  
Abstract: EGF receptor (EGFR)-targeted monoclonal antibodies are effective in a subset of metastatic colorectal cancers. Inevitably, all patients develop resistance, which occurs through emergence of KRAS mutations in approximately 50% of the cases. We show that amplification of the MET proto-oncogene is associated with acquired resistance in tumors that do not develop KRAS mutations during anti-EGFR therapy. Amplification of the MET locus was present in circulating tumor DNA before relapse was clinically evident. Functional studies show that MET activation confers resistance to anti-EGFR therapy both in vitro and in vivo. Notably, in patient-derived colorectal cancer xenografts, MET amplification correlated with resistance to EGFR blockade, which could be overcome by MET kinase inhibitors. These results highlight the role of MET in mediating primary and secondary resistance to anti-EGFR therapies in colorectal cancer and encourage the use of MET inhibitors in patients displaying resistance as a result of MET amplification.
Notes:
Emily Crowley, Federica Di Nicolantonio, Fotios Loupakis, Alberto Bardelli (2013)  Liquid biopsy: monitoring cancer-genetics in the blood.   Nature reviews. Clinical oncology Jul  
Abstract: Cancer is associated with mutated genes, and analysis of tumour-linked genetic alterations is increasingly used for diagnostic, prognostic and treatment purposes. The genetic profile of solid tumours is currently obtained from surgical or biopsy specimens; however, the latter procedure cannot always be performed routinely owing to its invasive nature. Information acquired from a single biopsy provides a spatially and temporally limited snap-shot of a tumour and might fail to reflect its heterogeneity. Tumour cells release circulating free DNA (cfDNA) into the blood, but the majority of circulating DNA is often not of cancerous origin, and detection of cancer-associated alleles in the blood has long been impossible to achieve. Technological advances have overcome these restrictions, making it possible to identify both genetic and epigenetic aberrations. A liquid biopsy, or blood sample, can provide the genetic landscape of all cancerous lesions (primary and metastases) as well as offering the opportunity to systematically track genomic evolution. This Review will explore how tumour-associated mutations detectable in the blood can be used in the clinic after diagnosis, including the assessment of prognosis, early detection of disease recurrence, and as surrogates for traditional biopsies with the purpose of predicting response to treatments and the development of acquired resistance.
Notes:
Davide Zecchin, Sabrina Arena, Miriam Martini, Francesco Sassi, Alberto Pisacane, Federica Di Nicolantonio, Alberto Bardelli (2013)  Modeling tumor progression by the sequential introduction of genetic alterations into the genome of human normal cells.   Hum Mutat 34: 2. 330-337 Feb  
Abstract: Cancer genomes display a complex blend of genetic lesions affecting oncogenes and tumor suppressor genes. Multiple modeling approaches indicate that 5-15 driver oncogenic events are required to achieve tumor progression in common epithelial cancers. In vitro, a lower number (2-3) of events is typically sufficient to achieve full transformation. We developed cellular models that closely resemble the occurrence of multiple genetic lesions to understand their role in tumor progression. Homologous recombination and transcriptional downregulation were used to recapitulate the co-occurrence of driver mutations targeting oncogenes and inactivation of tumor suppressor genes in human nontransformed epithelial cells. Knockdown of the tumor suppressor genes PTEN or RB1 was combined with mutagenic activation of individual oncogenes (EGFR, KRAS, BRAF, or PIK3CA), thus generating a combinatorial model. The simultaneous presence of oncogenic and tumor suppressive events resulted in distinct biochemical properties and anchorage-independent growth abilities. Notably, however, we found that even when up to four individual alterations were concomitantly present they were not sufficient to fully transform the target cells. Our results suggest that the close recapitulation of cancer lesions in not-transformed cells is essential to unveil their oncogenic potential and raise questions concerning the minimal requirements for neoplastic transformation of epithelial cells.
Notes:
Emanuele Valtorta, Sandra Misale, Andrea Sartore-Bianchi, Iris D Nagtegaal, François Paraf, Calogero Lauricella, Valentina Dimartino, Sebastijan Hobor, Bart Jacobs, Cristiana Ercolani, Simona Lamba, Elisa Scala, Silvio Veronese, Pierre Laurent-Puig, Salvatore Siena, Sabine Tejpar, Marcella Mottolese, Cornelis J A Punt, Marcello Gambacorta, Alberto Bardelli, Federica Di Nicolantonio (2013)  KRAS gene amplification in colorectal cancer and impact on response to EGFR-targeted therapy.   Int J Cancer Feb  
Abstract: KRAS mutations are the most common oncogenic event in colorectal cancer (CRC) progression and their occurrence is associated with lack of response to anti epidermal growth factor receptor (EGFR) targeted therapies. Using preclinical models and patients' samples we recently reported that the emergence of KRAS mutations but also KRAS amplification is associated with acquired resistance to the EGFR inhibitors cetuximab or panitumumab. We reasoned that KRAS amplification may also be responsible for primary resistance to these agents. Furthermore, while the prevalence of KRAS mutations has been well established in CRC, little is known about the frequency of KRAS amplification in large CRC series. We performed a screening of 1,039 CRC samples to assess the prevalence of KRAS amplification in this tumor type and further evaluated the role of this genetic alteration on the sensitivity to anti EGFR therapies. We detected KRAS amplification in 7/1,039 (0.67%) and 1/102 evaluable CRC specimens and cell lines, respectively. KRAS amplification was mutually exclusive with KRAS mutations. Tumors or cell lines harboring this genetic lesion are not responsive to anti-EGFR inhibitors. Although KRAS amplification is an infrequent event in CRC, it might be responsible for precluding response to anti-EGFR treatment in a small proportion of patients.
Notes:
2012
Alessia Bottos, Miriam Martini, Federica Di Nicolantonio, Valentina Comunanza, Federica Maione, Alberto Minassi, Giovanni Appendino, Federico Bussolino, Alberto Bardelli (2012)  Targeting oncogenic serine/threonine-protein kinase BRAF in cancer cells inhibits angiogenesis and abrogates hypoxia.   Proc Natl Acad Sci U S A 109: 6. E353-E359 Feb  
Abstract: Carcinomas are comprised of transformed epithelial cells that are supported in their growth by a dedicated neovasculature. How the genetic milieu of the epithelial compartment influences tumor angiogenesis is largely unexplored. Drugs targeted to mutant cancer genes may act not only on tumor cells but also, directly or indirectly, on the surrounding stroma. We investigated the role of the BRAF(V600E) oncogene in tumor/vessel crosstalk and analyzed the effect of the BRAF inhibitor PLX4720 on tumor angiogenesis. Knock-in of the BRAF(V600E) allele into the genome of human epithelial cells triggered their angiogenic response. In cancer cells harboring oncogenic BRAF, the inhibitor PLX4720 switches off the ERK pathway and inhibits the expression of proangiogenic molecules. In tumor xenografts harboring the BRAF(V600E), PLX4720 extensively modifies the vascular network causing abrogation of hypoxia. Overall, our results provide a functional link between oncogenic BRAF and angiogenesis. Furthermore, they indicate how the tumor vasculature can be "indirectly" besieged through targeting of a genetic lesion to which the cancer cells are addicted.
Notes:
Anirudh Prahallad, Chong Sun, Sidong Huang, Federica Di Nicolantonio, Ramon Salazar, Davide Zecchin, Roderick L Beijersbergen, Alberto Bardelli, René Bernards (2012)  Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR.   Nature 483: 7387. 100-103 Mar  
Abstract: Inhibition of the BRAF(V600E) oncoprotein by the small-molecule drug PLX4032 (vemurafenib) is highly effective in the treatment of melanoma. However, colon cancer patients harbouring the same BRAF(V600E) oncogenic lesion have poor prognosis and show only a very limited response to this drug. To investigate the cause of the limited therapeutic effect of PLX4032 in BRAF(V600E) mutant colon tumours, here we performed an RNA-interference-based genetic screen in human cells to search for kinases whose knockdown synergizes with BRAF(V600E) inhibition. We report that blockade of the epidermal growth factor receptor (EGFR) shows strong synergy with BRAF(V600E) inhibition. We find in multiple BRAF(V600E) mutant colon cancers that inhibition of EGFR by the antibody drug cetuximab or the small-molecule drugs gefitinib or erlotinib is strongly synergistic with BRAF(V600E) inhibition, both in vitro and in vivo. Mechanistically, we find that BRAF(V600E) inhibition causes a rapid feedback activation of EGFR, which supports continued proliferation in the presence of BRAF(V600E) inhibition. Melanoma cells express low levels of EGFR and are therefore not subject to this feedback activation. Consistent with this, we find that ectopic expression of EGFR in melanoma cells is sufficient to cause resistance to PLX4032. Our data suggest that BRAF(V600E) mutant colon cancers (approximately 8-10% of all colon cancers), for which there are currently no targeted treatment options available, might benefit from combination therapy consisting of BRAF and EGFR inhibitors.
Notes:
Giorgia Migliardi, Francesco Sassi, Davide Torti, Francesco Galimi, Eugenia R Zanella, Michela Buscarino, Dario Ribero, Andrea Muratore, Paolo Massucco, Alberto Pisacane, Mauro Risio, Lorenzo Capussotti, Silvia Marsoni, Federica Di Nicolantonio, Alberto Bardelli, Paolo M Comoglio, Livio Trusolino, Andrea Bertotti (2012)  Inhibition of MEK and PI3K/mTOR Suppresses Tumor Growth but Does Not Cause Tumor Regression in Patient-Derived Xenografts of RAS-Mutant Colorectal Carcinomas.   Clin Cancer Res 18: 9. 2515-2525 May  
Abstract: PURPOSE: Gene mutations along the Ras pathway (KRAS, NRAS, BRAF, PIK3CA) occur in approximately 50% of colorectal cancers (CRC) and correlate with poor response to anti-EGF receptor (EGFR) therapies. We assessed the effects of mitogen-activated protein (MAP)/extracellular signal-regulated kinase (ERK) kinase (MEK) and phosphoinositide 3-kinase (PI3K)/mTOR inhibitors, which neutralize the major Ras effectors, in patient-derived xenografts from RAS/RAF/PIK3CA-mutant metastatic CRCs (mCRC). EXPERIMENTAL DESIGN: Forty mCRC specimens harboring KRAS, NRAS, BRAF, and/or PIK3CA mutations were implanted in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. Each xenograft was expanded into four treatment arms: placebo, the MEK inhibitor AZD6244, the PI3K/mTOR inhibitor, BEZ235, or AZD6244 + BEZ235. Cases initially treated with placebo crossed over to AZD6244, BEZ235, and the anti-EGFR monoclonal antibody cetuximab. RESULTS: At the 3-week evaluation time point, cotreatment of established tumors with AZD6244 + BEZ235 induced disease stabilization in the majority of cases (70%) but did not lead to overt tumor regression. Monotherapy was less effective, with BEZ235 displaying higher activity than AZD6244 (disease control rates, DCRs: AZD6244, 27.5%; BEZ235, 42.5%). Triple therapy with cetuximab provided further advantage (DCR, 88%). The extent of disease control declined at the 6-week evaluation time point (DCRs: AZD6244, 13.9%; BEZ235, 16.2%; AZD6244 + BEZ235, 34%). Cross-analysis of mice harboring xenografts from the same original tumor and treated with each of the different modalities revealed subgroups with preferential sensitivity to AZD6244 (12.5%), BEZ235 (35%), or AZD6244 + BEZ235 (42.5%); another subgroup (10%) showed equivalent response to any treatment. CONCLUSIONS: The prevalent growth-suppressive effects produced by MEK and PI3K/mTOR inhibition suggest that this strategy may retard disease progression in patients. However, data offer cautionary evidence against the occurrence of durable responses. Clin Cancer Res; 18(9); 2515-25. ©2012 AACR.
Notes:
Sandra Misale, Rona Yaeger, Sebastijan Hobor, Elisa Scala, Manickam Janakiraman, David Liska, Emanuele Valtorta, Roberta Schiavo, Michela Buscarino, Giulia Siravegna, Katia Bencardino, Andrea Cercek, Chin-Tung Chen, Silvio Veronese, Carlo Zanon, Andrea Sartore-Bianchi, Marcello Gambacorta, Margherita Gallicchio, Efsevia Vakiani, Valentina Boscaro, Enzo Medico, Martin Weiser, Salvatore Siena, Federica Di Nicolantonio, David Solit, Alberto Bardelli (2012)  Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer.   Nature 486: 7404. 532-536 Jun  
Abstract: A main limitation of therapies that selectively target kinase signalling pathways is the emergence of secondary drug resistance. Cetuximab, a monoclonal antibody that binds the extracellular domain of epidermal growth factor receptor (EGFR), is effective in a subset of KRAS wild-type metastatic colorectal cancers. After an initial response, secondary resistance invariably ensues, thereby limiting the clinical benefit of this drug. The molecular bases of secondary resistance to cetuximab in colorectal cancer are poorly understood. Here we show that molecular alterations (in most instances point mutations) of KRAS are causally associated with the onset of acquired resistance to anti-EGFR treatment in colorectal cancers. Expression of mutant KRAS under the control of its endogenous gene promoter was sufficient to confer cetuximab resistance, but resistant cells remained sensitive to combinatorial inhibition of EGFR and mitogen-activated protein-kinase kinase (MEK). Analysis of metastases from patients who developed resistance to cetuximab or panitumumab showed the emergence of KRAS amplification in one sample and acquisition of secondary KRAS mutations in 60% (6 out of 10) of the cases. KRAS mutant alleles were detectable in the blood of cetuximab-treated patients as early as 10 months before radiographic documentation of disease progression. In summary, the results identify KRAS mutations as frequent drivers of acquired resistance to cetuximab in colorectal cancers, indicate that the emergence of KRAS mutant clones can be detected non-invasively months before radiographic progression and suggest early initiation of a MEK inhibitor as a rational strategy for delaying or reversing drug resistance.
Notes:
2011
Davide Zecchin, Federica Di Nicolantonio (2011)  Transfection and DNA-mediated gene transfer.   Methods Mol Biol 731: 435-450  
Abstract: The advent of recent technologies such as gene expression microarrays and high-throughput sequencing methods has allowed for unveiling the molecular complexity of cancer. However, compared to the genomic discovery stage, the functional characterization of genes that have been found altered (by somatic mutations, rearrangements, or copy number variations) or differentially regulated at the expression level is still lagging behind. In the future, it is anticipated that efforts would be aimed at addressing the impact of such genes on several cancer traits, including tumor formation, dissemination, and response to therapies. These studies would likely have to rely on introducing the gene(s) of interest (in its -wild-type or altered version) in cellular models. We describe here a number of techniques to introduce nucleic acids into eukaryotic cells, ranging from conventional plasmid transfection to lentiviral -transduction and adeno-associated viral (AAV)-mediated DNA transfer.
Notes:
Andrea Bertotti, Giorgia Migliardi, Francesco Galimi, Francesco Sassi, Davide Torti, Claudio Isella, Davide Corà, Federica Di Nicolantonio, Michela Buscarino, Consalvo Petti, Dario Ribero, Nadia Russolillo, Andrea Muratore, Paolo Massucco, Alberto Pisacane, Luca Molinaro, Emanuele Valtorta, Andrea Sartore-Bianchi, Mauro Risio, Lorenzo Capussotti, Marcello Gambacorta, Salvatore Siena, Enzo Medico, Anna Sapino, Silvia Marsoni, Paolo M Comoglio, Alberto Bardelli, Livio Trusolino (2011)  A Molecularly Annotated Platform of Patient-Derived Xenografts ("Xenopatients") Identifies HER2 as an Effective Therapeutic Target in Cetuximab-Resistant Colorectal Cancer.   Cancer Discov 1: 6. 508-523  
Abstract: Only a fraction of patients with metastatic colorectal cancer receive clinical benefit from therapy with anti-epidermal growth factor receptor (EGFR) antibodies, which calls for the identification of novel biomarkers for better personalized medicine. We produced large xenograft cohorts from 85 patient-derived, genetically characterized metastatic colorectal cancer samples ("xenopatients") to discover novel determinants of therapeutic response and new oncoprotein targets. Serially passaged tumors retained the morphologic and genomic features of their original counterparts. A validation trial confirmed the robustness of this approach: xenopatients responded to the anti-EGFR antibody cetuximab with rates and extents analogous to those observed in the clinic and could be prospectively stratified as responders or nonresponders on the basis of several predictive biomarkers. Genotype-response correlations indicated HER2 amplification specifically in a subset of cetuximab-resistant, KRAS/NRAS/BRAF/PIK3CA wild-type cases. Importantly, HER2 amplification was also enriched in clinically nonresponsive KRAS wild-type patients. A proof-of-concept, multiarm study in HER2-amplified xenopatients revealed that the combined inhibition of HER2 and EGFR induced overt, long-lasting tumor regression. Our results suggest promising therapeutic opportunities in cetuximab-resistant patients with metastatic colorectal cancer, whose medical treatment in the chemorefractory setting remains an unmet clinical need.
Notes:
Francesca Molinari, Lara Felicioni, Michela Buscarino, Sara De Dosso, Fiamma Buttitta, Sara Malatesta, Alessandra Movilia, Marco Luoni, Renzo Boldorini, Oscar Alabiso, Salvatore Girlando, Barbara Soini, Alessandra Spitale, Federica Di Nicolantonio, Piercarlo Saletti, Stefano Crippa, Luca Mazzucchelli, Antonio Marchetti, Alberto Bardelli, Milo Frattini (2011)  Increased detection sensitivity for KRAS mutations enhances the prediction of anti-EGFR monoclonal antibody resistance in metastatic colorectal cancer.   Clin Cancer Res 17: 14. 4901-4914 Jul  
Abstract: KRAS mutations represent the main cause of resistance to anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (MoAbs) in metastatic colorectal cancer (mCRC). We evaluated whether highly sensitive methods for KRAS investigation improve the accuracy of predictions of anti-EGFR MoAbs efficacy.
Notes:
Roberta Rosa, Davide Melisi, Vincenzo Damiano, Roberto Bianco, Sonia Garofalo, Teresa Gelardi, Sudhir Agrawal, Federica Di Nicolantonio, Aldo Scarpa, Alberto Bardelli, Giampaolo Tortora (2011)  Toll-like receptor 9 agonist IMO cooperates with cetuximab in K-ras mutant colorectal and pancreatic cancers.   Clin Cancer Res 17: 20. 6531-6541 Oct  
Abstract: K-Ras somatic mutations are a strong predictive biomarker for resistance to epidermal growth factor receptor (EGFR) inhibitors in patients with colorectal and pancreatic cancer. We previously showed that the novel Toll-like receptor 9 (TLR9) agonist immunomodulatory oligonucleotide (IMO) has a strong in vivo activity in colorectal cancer models by interfering with EGFR-related signaling and synergizing with the anti-EGFR monoclonal antibody cetuximab.
Notes:
2010
Andrea Sartore-Bianchi, Katia Bencardino, Federica Di Nicolantonio, Federico Pozzi, Chiara Funaioli, Valentina Gambi, Sabrina Arena, Miriam Martini, Simona Lamba, Andrea Cassingena, Roberta Schiavo, Alberto Bardelli, Salvatore Siena (2010)  Integrated molecular dissection of the epidermal growth factor receptor (EFGR) oncogenic pathway to predict response to EGFR-targeted monoclonal antibodies in metastatic colorectal cancer.   Target Oncol 5: 1. 19-28 Mar  
Abstract: The introduction of KRAS testing as a diagnostic tool to select patients for epidermal growth factor receptor (EGFR)-targeted cetuximab- or panitumumab-based therapies for metastatic colorectal cancer is widely regarded as a key advance in the field of personalized cancer medicine. Oncologists are now facing emerging issues in the treatment of metastatic colorectal cancer, including: (i) the identification of additional genetic determinants of primary resistance to EGFR-targeted therapy for further improving selection of patients; (ii) the explanation of rare cases of patients carrying KRAS-mutated tumors who have been reported to respond to either cetuximab or panitumumab and (iii) the discovery of mechanisms of secondary resistance to anti-EGFR antibody therapies. Here we discuss the potential role of comprehensive dissection of the key oncogenic nodes in the EGFR signaling cascade to predict resistance and sensitivity to EGFR monoclonal antibodies in metastatic colorectal cancer. Current data suggest that, together with KRAS mutations, the evaluation of BRAF and PIK3CA/PTEN alterations could also be useful for selecting patients with reduced chance to benefit from EGFR-targeted therapy. Furthermore, measuring EGFR gene copy number also appears relevant to positively identify responders. Up until now, each of these markers has been mainly assessed as a single event, often in retrospective analyses and patients' series. As these molecular alterations display overlapping patterns of occurrence, this adds considerable complexity to the drawing of an algorithm suitable for clinical decision-making. We suggest that in the near future comprehensive molecular analysis of the entire oncogenic pathway triggered by the EGFR should be performed, thus enhancing the prediction ability of individual markers.
Notes:
Federica Di Nicolantonio, Sabrina Arena, Josep Tabernero, Stefano Grosso, Francesca Molinari, Teresa Macarulla, Mariangela Russo, Carlotta Cancelliere, Davide Zecchin, Luca Mazzucchelli, Takehiko Sasazuki, Senji Shirasawa, Massimo Geuna, Milo Frattini, José Baselga, Margherita Gallicchio, Stefano Biffo, Alberto Bardelli (2010)  Deregulation of the PI3K and KRAS signaling pathways in human cancer cells determines their response to everolimus.   J Clin Invest 120: 8. 2858-2866 Aug  
Abstract: Personalized cancer medicine is based on the concept that targeted therapies are effective on subsets of patients whose tumors carry specific molecular alterations. Several mammalian target of rapamycin (mTOR) inhibitors are in preclinical or clinical trials for cancers, but the molecular basis of sensitivity or resistance to these inhibitors among patients is largely unknown. Here we have identified oncogenic variants of phosphoinositide-3-kinase, catalytic, alpha polypeptide (PIK3CA) and KRAS as determinants of response to the mTOR inhibitor everolimus. Human cancer cells carrying alterations in the PI3K pathway were responsive to everolimus, both in vitro and in vivo, except when KRAS mutations occurred concomitantly or were exogenously introduced. In human cancer cells with mutations in both PIK3CA and KRAS, genetic ablation of mutant KRAS reinstated response to the drug. Consistent with these data, PIK3CA mutant cells, but not KRAS mutant cells, displayed everolimus-sensitive translation. Importantly, in a cohort of metastatic cancer patients, the presence of oncogenic KRAS mutations was associated with lack of benefit after everolimus therapy. Thus, our results demonstrate that alterations in the KRAS and PIK3CA genes may represent biomarkers to optimize treatment of patients with mTOR inhibitors.
Notes:
A Sartore-Bianchi, K Bencardino, A Cassingena, F Venturini, C Funaioli, T Cipani, A Amatu, L Pietrogiovanna, R Schiavo, F Di Nicolantonio, S Artale, A Bardelli, S Siena (2010)  Therapeutic implications of resistance to molecular therapies in metastatic colorectal cancer.   Cancer Treat Rev 36 Suppl 3: S1-S5 Nov  
Abstract: Metastatic colorectal cancer (mCRC) patients carrying KRAS mutated tumors do not benefit from epidermal growth factor receptor (EGFR)-targeted cetuximab- or panitumumab-based therapies. Indeed, the mutational status of KRAS is currently a validated predictive biomarker employed to select mCRC patients for EGFR targeted drugs. When patients fail standard 5-fluorouracil-, oxaliplatin-, irinotecan- and bevacizumab-based therapies, EGFR-targeted salvage therapy can be prescribed only for those individuals with KRAS wild-type cancer. Thus, clinicians are now facing the urgent issue of better understanding the biology of KRAS mutant disease, in order to devise novel effective therapies in such defined genetic setting. In addition to KRAS, recent data point out that BRAF and PIK3CA exon 20 mutations hamper response to EGFR-targeted treatment in mCRC, potentially excluding from treatment also patients with these molecular alterations in their tumor. This review will focus on current knowledge regarding the molecular landscape of mCRC including and beyond KRAS, and will summarize novel rationally-developed combinatorial regimens that are being evaluated in early clinical trials.
Notes:
2009
Alessandro Palmioli, Elena Sacco, Cristina Airoldi, Federica Di Nicolantonio, Annalisa D'Urzo, Senji Shirasawa, Takehiko Sasazuki, Alessandro Di Domizio, Luca De Gioia, Enzo Martegani, Alberto Bardelli, Francesco Peri, Marco Vanoni (2009)  Selective cytotoxicity of a bicyclic Ras inhibitor in cancer cells expressing K-Ras(G13D).   Biochem Biophys Res Commun 386: 4. 593-597 Sep  
Abstract: Mutation of RAS genes is a critical event in the pathogenesis of different human tumors and in some developmental disorders. Here we present an arabinose-derived bicyclic compound displaying selective cytotoxicity in human colorectal cancer cells expressing K-Ras(G13D), that shows high intrinsic nucleotide exchange rate. We characterize binding of bicyclic compounds by docking and NMR experiments and their inhibitory activity on GEF-mediated nucleotide exchange on wild-type and mutant Ras proteins. We demonstrate that the in vitro inhibition of Ras nucleotide exchange depends on the molar ratio between Ras and its GEF activator, suggesting that the observed in vivo selective effect may depend on biochemical parameters and actual intracellular concentration of the Ras protein and its regulators.
Notes:
Andrea Sartore-Bianchi, Miriam Martini, Francesca Molinari, Silvio Veronese, Michele Nichelatti, Salvatore Artale, Federica Di Nicolantonio, Piercarlo Saletti, Sara De Dosso, Luca Mazzucchelli, Milo Frattini, Salvatore Siena, Alberto Bardelli (2009)  PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies.   Cancer Res 69: 5. 1851-1857 Mar  
Abstract: The monoclonal antibodies (moAb) panitumumab and cetuximab target the epidermal growth factor receptor (EGFR) and have proven valuable for the treatment of metastatic colorectal cancer (mCRC). EGFR-mediated signaling involves two main intracellular cascades: on one side KRAS activates BRAF, which in turn triggers the mitogen-activated protein kinases. On the other, membrane localization of the lipid kinase PIK3CA counteracts PTEN and promotes AKT1 phosphorylation, thereby activating a parallel intracellular axis. Constitutive activation of KRAS bypasses the corresponding signaling cascade and, accordingly, patients with mCRC bearing KRAS mutations are clinically resistant to therapy with panitumumab or cetuximab. We hypothesized that mutations activating PIK3CA could also preclude responsiveness to EGFR-targeted moAbs through a similar mechanism. Here, we present the mutational analysis of PIK3CA and KRAS and evaluation of the PTEN protein status in a cohort of 110 patients with mCRC treated with anti-EGFR moAbs. We observed 15 (13.6%) PIK3CA and 32 (29.0%) KRAS mutations. PIK3CA mutations were significantly associated with clinical resistance to panitumumab or cetuximab; none of the mutated patients achieved objective response (P = 0.038). When only KRAS wild-type tumors were analyzed, the statistical correlation was stronger (P = 0.016). Patients with PIK3CA mutations displayed a worse clinical outcome also in terms of progression-free survival (P = 0.035). Our data indicate that PIK3CA mutations can independently hamper the therapeutic response to panitumumab or cetuximab in mCRC. When the molecular status of the PIK3CA/PTEN and KRAS pathways are concomitantly ascertained, up to 70% of mCRC patients unlikely to respond to EGFR moAbs can be identified.
Notes:
Salvatore Siena, Andrea Sartore-Bianchi, Federica Di Nicolantonio, Julia Balfour, Alberto Bardelli (2009)  Biomarkers predicting clinical outcome of epidermal growth factor receptor-targeted therapy in metastatic colorectal cancer.   J Natl Cancer Inst 101: 19. 1308-1324 Oct  
Abstract: The monoclonal antibodies panitumumab and cetuximab that target the epidermal growth factor receptor (EGFR) have expanded the range of treatment options for metastatic colorectal cancer. Initial evaluation of these agents as monotherapy in patients with EGFR-expressing chemotherapy-refractory tumors yielded response rates of approximately 10%. The realization that detection of positive EGFR expression by immunostaining does not reliably predict clinical outcome of EGFR-targeted treatment has led to an intense search for alternative predictive biomarkers. Oncogenic activation of signaling pathways downstream of the EGFR, such as mutation of KRAS, BRAF, or PIK3CA oncogenes, or inactivation of the PTEN tumor suppressor gene is central to the progression of colorectal cancer. Tumor KRAS mutations, which may be present in 35%-45% of patients with colorectal cancer, have emerged as an important predictive marker of resistance to panitumumab or cetuximab treatment. In addition, among colorectal tumors carrying wild-type KRAS, mutation of BRAF or PIK3CA or loss of PTEN expression may be associated with resistance to EGFR-targeted monoclonal antibody treatment, although these additional biomarkers require further validation before incorporation into clinical practice. Additional knowledge of the molecular basis for sensitivity or resistance to EGFR-targeted monoclonal antibodies will allow the development of new treatment algorithms to identify patients who are most likely to respond to treatment and could also provide rationale for combining therapies to overcome primary resistance. The use of KRAS mutations as a selection biomarker for anti-EGFR monoclonal antibody (eg, panitumumab or cetuximab) treatment is the first major step toward individualized treatment for patients with metastatic colorectal cancer.
Notes:
Andrea Sartore-Bianchi, Federica Di Nicolantonio, Michele Nichelatti, Francesca Molinari, Sara De Dosso, Piercarlo Saletti, Miriam Martini, Tiziana Cipani, Giovanna Marrapese, Luca Mazzucchelli, Simona Lamba, Silvio Veronese, Milo Frattini, Alberto Bardelli, Salvatore Siena (2009)  Multi-determinants analysis of molecular alterations for predicting clinical benefit to EGFR-targeted monoclonal antibodies in colorectal cancer.   PLoS One 4: 10. 10  
Abstract: BACKGROUND: KRAS mutations occur in 35-45% of metastatic colorectal cancers (mCRC) and preclude responsiveness to EGFR-targeted therapy with cetuximab or panitumumab. However, less than 20% patients displaying wild-type KRAS tumors achieve objective response. Alterations in other effectors downstream of the EGFR, such as BRAF, and deregulation of the PIK3CA/PTEN pathway have independently been found to give rise to resistance. We present a comprehensive analysis of KRAS, BRAF, PIK3CA mutations, and PTEN expression in mCRC patients treated with cetuximab or panitumumab, with the aim of clarifying the relative contribution of these molecular alterations to resistance. METHODOLOGY/PRINCIPAL FINDINGS: We retrospectively analyzed objective tumor response, progression-free (PFS) and overall survival (OS) together with the mutational status of KRAS, BRAF, PIK3CA and expression of PTEN in 132 tumors from cetuximab or panitumumab treated mCRC patients. Among the 106 non-responsive patients, 74 (70%) had tumors with at least one molecular alteration in the four markers. The probability of response was 51% (22/43) among patients with no alterations, 4% (2/47) among patients with 1 alteration, and 0% (0/24) for patients with > or =2 alterations (p<0.0001). Accordingly, PFS and OS were increasingly worse for patients with tumors harboring none, 1, or > or =2 molecular alteration(s) (p<0.001). CONCLUSIONS/SIGNIFICANCE: When expression of PTEN and mutations of KRAS, BRAF and PIK3CA are concomitantly ascertained, up to 70% of mCRC patients unlikely to respond to anti-EGFR therapies can be identified. We propose to define as 'quadruple negative', the CRCs lacking alterations in KRAS, BRAF, PTEN and PIK3CA. Comprehensive molecular dissection of the EGFR signaling pathways should be considered to select mCRC patients for cetuximab- or panitumumab-based therapies.
Notes:
2008
Federica Di Nicolantonio, Sabrina Arena, Margherita Gallicchio, Davide Zecchin, Miriam Martini, Simona Emilia Flonta, Giulia Maria Stella, Simona Lamba, Carlotta Cancelliere, Mariangela Russo, Massimo Geuna, Giovanni Appendino, Roberto Fantozzi, Enzo Medico, Alberto Bardelli (2008)  Replacement of normal with mutant alleles in the genome of normal human cells unveils mutation-specific drug responses.   Proc Natl Acad Sci U S A 105: 52. 20864-20869 Dec  
Abstract: Mutations in oncogenes and tumor suppressor genes are responsible for tumorigenesis and represent favored therapeutic targets in oncology. We exploited homologous recombination to knock-in individual cancer mutations in the genome of nontransformed human cells. Sequential introduction of multiple mutations was also achieved, demonstrating the potential of this strategy to construct tumor progression models. Knock-in cells displayed allele-specific activation of signaling pathways and mutation-specific phenotypes different from those obtainable by ectopic oncogene expression. Profiling of a library of pharmacological agents on the mutated cells showed striking sensitivity or resistance phenotypes to pathway-targeted drugs, often matching those of tumor cells carrying equivalent cancer mutations. Thus, knock-in of single or multiple cancer alleles provides a pharmacogenomic platform for the rational design of targeted therapies.
Notes:
Federica Di Nicolantonio, Miriam Martini, Francesca Molinari, Andrea Sartore-Bianchi, Sabrina Arena, Piercarlo Saletti, Sara De Dosso, Luca Mazzucchelli, Milo Frattini, Salvatore Siena, Alberto Bardelli (2008)  Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer.   J Clin Oncol 26: 35. 5705-5712 Dec  
Abstract: PURPOSE Cetuximab or panitumumab are effective in 10% to 20% unselected metastatic colorectal cancer (CRC) patients. KRAS mutations account for approximately 30% to 40% patients who are not responsive. The serine-threonine kinase BRAF is the principal effector of KRAS. We hypothesized that, in KRAS wild-type patients, BRAF mutations could have a predictive/prognostic value. PATIENTS AND METHODS We retrospectively analyzed objective tumor responses, time to progression, overall survival (OS), and the mutational status of KRAS and BRAF in 113 tumors from cetuximab- or panitumumab-treated metastatic CRC patients. The effect of the BRAF V600E mutation on cetuximab or panitumumab response was also assessed using cellular models of CRC. Results KRAS mutations were present in 30% of the patients and were associated with resistance to cetuximab or panitumumab (P = .011). The BRAF V600E mutation was detected in 11 of 79 patients who had wild-type KRAS. None of the BRAF-mutated patients responded to treatment, whereas none of the responders carried BRAF mutations (P = .029). BRAF-mutated patients had significantly shorter progression-free survival (P = .011) and OS (P < .0001) than wild-type patients. In CRC cells, the introduction of BRAF V600E allele impaired the therapeutic effect of cetuximab or panitumumab. Treatment with the BRAF inhibitor sorafenib restored sensitivity to panitumumab or cetuximab of CRC cells carrying the V600E allele. CONCLUSION BRAF wild-type is required for response to panitumumab or cetuximab and could be used to select patients who are eligible for the treatment. Double-hit therapies aimed at simultaneous inhibition of epidermal growth factor receptor and BRAF warrant exploration in CRC patients carrying the V600E oncogenic mutation.
Notes:
2007
Silvia Benvenuti, Andrea Sartore-Bianchi, Federica Di Nicolantonio, Carlo Zanon, Mauro Moroni, Silvio Veronese, Salvatore Siena, Alberto Bardelli (2007)  Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti-epidermal growth factor receptor antibody therapies.   Cancer Res 67: 6. 2643-2648 Mar  
Abstract: Monoclonal antibodies (mAbs) against the extracellular domain of the epidermal growth factor receptor (EGFR) have been introduced for the treatment of metastatic colorectal cancer (mCRC). We have reported recently that increased copy number of the EGFR can predict response to anti-EGFR mAbs and that patients might be selected for treatment based on EGFR copy number. Here, we show that mutations activating the RAS/RAF signaling pathway are also predictive and prognostic indicators in mCRC patients, being inversely correlated with response to anti-EGFR mAbs. In cellular models of CRCs, activation of the RAS signaling pathway by introduction of an activated K-RAS allele (Gly(12)Val) impairs the therapeutic effect of anti-EGFR mAbs. In cancer cells carrying constitutively active RAS, the pharmacologic inhibition of the mitogen-activated protein kinase (MAPK) signaling cascade improves anti-EGFR treatment based on mAbs. These results have implications for the identification of patients who are likely to respond to anti-EGFR treatment. They also provide the rationale for combination therapies, targeted simultaneously to the EGFR and RAS/RAF/MAPK signaling pathways in CRC patients.
Notes:
2006
Federica Di Nicolantonio, Alberto Bardelli (2006)  Kinase mutations in cancer: chinks in the enemy's armour?   Curr Opin Oncol 18: 1. 69-76 Jan  
Abstract: PURPOSE OF REVIEW: Over the past few years, a revolution has transformed the oncology field. This revolution is characterized by two main features. The first is the introduction of the concept of individualized cancer therapy. The second is the development of drugs targeting molecules selectively altered in tumours. This review analyses these aspects by looking at the role that altered kinases and their inhibitors have played in this historical process. RECENT FINDINGS: Tumour progression is the result of the sequential accumulation of mutations in genes monitoring the rates of cell birth and cell death. The molecular profiling of cancers has shown that protein and lipid kinases are frequently altered in tumour cells. In most cases, these alterations translate in constitutively active proteins, which are amenable of therapeutic targeting. Intriguingly, even 'established' cancer cells remain somewhat 'addicted' to the deregulated activity of mutated kinases. This feature appears to be the basis for the ability of kinase inhibitors in controlling the development of a number of cancers. The therapeutic efficacy of kinase inhibitors is impaired by the emergence of tumour cells carrying 'resistance' mutations. SUMMARY: Many oncogenes are mutated kinase genes. In most cases, the mutations result in the constitutive activation of the affected kinase that can be pharmacologically inhibited. Unfortunately, upon treatment with kinase inhibitors, resistant clones develop rapidly, impairing their therapeutic effect. Strategies to overcome resistance are discussed as well as the possibility to target kinases regulating cancer stem cells.
Notes:
Pradeep Bhandari, Adrian C Bateman, Raj L Mehta, Bernard Sf Stacey, Penny Johnson, Ian A Cree, Federica Di Nicolantonio, Praful Patel (2006)  Prognostic significance of cyclooxygenase-2 (COX-2) expression in patients with surgically resectable adenocarcinoma of the oesophagus.   BMC Cancer 6: 05  
Abstract: BACKGROUND: COX-2 expression in tumour cells has been associated with poor prognosis in gastrointestinal and non-gastrointestinal cancers. The aim of our study was to test the hypothesis that higher levels of COX-2 expression are prognostically related to poor clinico-pathologic features in adenocarcinoma of the oesophagus. METHODS: We reviewed the records of 100 consecutive patients undergoing resection for adenocarcinoma of the oesophagus to collect data on T-stage, N-stage, tumour recurrence and survival. T & N-stage was further confirmed by histological examination. COX-2 protein expression was assessed by immunohistochemistry in all patients and COX-2 m-RNA expression was measured by quantitative RT-PCR in a small group of patients. RESULTS: Higher levels of COX-2 expression were associated with higher T stage (p = 0.008), higher N stage (p = 0.049), increased risk of tumour recurrence (p = 0.01) and poor survival (p = <0.001). A COX-2 score of >200 was associated with a median survival of 10 months compared to 26 months with a score of <200 (p = <0.001). CONCLUSION: Higher levels of COX-2 expression are associated with poor clinico-pathologic features and poor survival in patients with oesophageal adenocarcinoma.
Notes:
Louise A Knight, Federica Di Nicolantonio, Pauline A Whitehouse, Stuart J Mercer, Sanjay Sharma, Sharon Glaysher, John L Hungerford, Jeremy Hurren, Alan Lamont, Ian A Cree (2006)  The effect of imatinib mesylate (Glivec) on human tumor-derived cells.   Anticancer Drugs 17: 6. 649-655 Jul  
Abstract: Imatinib mesylate is a specific inhibitor of the Bcr-Abl protein tyrosine kinase that competes with ATP for its specific binding site in the kinase domain. It has activity against platelet-derived growth factor receptor alpha and beta (PDGFR-alpha and -beta), and c-kit, the receptor for stem cell factor. We have used a standardized ATP-tumor chemosensitivity assay and immunohistochemistry to determine the cytotoxicity of imatinib mesylate in tumor-derived cells from cutaneous and uveal melanoma, and ovarian carcinoma. Imatinib mesylate was tested at concentrations ranging from 2.0 to 0.0625 micromol/l alone and in combination with a cytotoxic drug (cisplatin, doxorubicin, paclitaxel or treosulfan). Imatinib mesylate showed low inhibition (IndexSUM>300) across the range of concentrations tested in this study, with few tumors exhibiting increasing inhibition with increased drug concentration. The median IC90 values for cutaneous and uveal melanoma and ovarian carcinoma were 13.2 micromol/l (4.0-294.3 micromol/l), 12.0 micromol/l (2.0-285.4 micromol/l) and 7.71 micromol/l (6.51-11.02 micromol/l), respectively. Imatinib mesylate potentiated the effect of different cytotoxics in 9% (5/54) of cases and had a negative effect in 13% (7/54) of cases, with no effect in the remainder. No correlation of effect was noted with c-kit, platelet-derived growth factor receptor-alpha or platelet-derived growth factor receptor-beta expression, assessed by immunohistochemistry. The signaling pathways mediated by activation of c-kit or platelet-derived growth factor receptor may act as antiapoptotic survival signals in some cancers and inhibition of these pathways may potentiate the activity of some cytotoxic drugs by inhibiting the survival signal. Growth inhibition, however, may reduce the efficacy of cytotoxic drugs, which tend to target proliferating cells preferentially, and clinical effects are therefore difficult to predict.
Notes:
Augusta Fernando, Sharon Glaysher, Mark Conroy, Marcin Pekalski, Jason Smith, Louise A Knight, Federica Di Nicolantonio, Ian A Cree (2006)  Effect of culture conditions on the chemosensitivity of ovarian cancer cell lines.   Anticancer Drugs 17: 8. 913-919 Sep  
Abstract: The aim of this study was to define the chemosensitivity profile of a series of human ovarian cancer cell lines representing the human primary ovarian tumours under altered culture conditions and to compare the results with those from tumour-derived cells. In this study, we used a standardized ATP-based tumour chemosensitivity assay to measure the activity of cytotoxics in the seven ovarian carcinoma cell lines and ovarian tumour-derived cells. The use of adherence-free polypropylene plates and a serum-free medium slowed down cell proliferation in all cell lines tested, mimicking the slow growth rate of solid tumours in this type of plastic. The seeding density was optimized for each cell line and was in the range of 2000-4000 cells/well. Heterogenous sensitivity to different cytotoxics was observed in the seven ovarian cancer cell lines tested in the ATP-based tumour chemosensitivity assay. The human ovarian carcinoma cell line, OVCA433, was found to be the most resistant cell line and 75% of the drugs showed an Index(SUM) above 300. Our results suggest that the use of appropriate culture conditions i.e. a serum-free culture environment, adherence-free growth and optimum seeding density can induce cell lines to behave more like tumour-derived cells in response to cytotoxic agents. On the basis of the comparison of chemosensitivity profiles of tumour-derived cells and cell lines derived from the corresponding tumour, a panel of cell lines can be selected. Such a panel could be used to screen and develop anticancer drugs.
Notes:
2005
Stuart J Mercer, Federica Di Nicolantonio, Louise A Knight, Francis G Gabriel, Pauline A Whitehouse, Sanjay Sharma, Augusta Fernando, Pradeep Bhandari, Shaw S Somers, Simon K Toh, Ian A Cree (2005)  Rapid up-regulation of cyclooxygenase-2 by 5-fluorouracil in human solid tumors.   Anticancer Drugs 16: 5. 495-500 Jun  
Abstract: Inhibition of cyclooxygenase (COX)-2 has been associated with reduced growth of malignant cells. Current therapy of gastrointestinal carcinomas involves the use of 5-fluorouracil (5-FU)-based chemotherapy and we have therefore studied the effect of this agent on the expression of COX-2. COX-2 expression was measured by quantitative RT-PCR in biopsies from a series of 14 esophageal carcinomas, six of which had paired samples taken before and after chemotherapy, and in tumor-derived cells exposed to 5-FU in vitro from a series of 44 tumors, including breast, ovarian, esophageal and colonic carcinomas. COX-2 expression was increased by exposure to 5-FU or 5-FU combination chemotherapy in all the tumor types studied, whether measured in biopsies taken before and after 5-FU-based chemotherapy (4-fold increase, p<0.015) or in primary cells exposed to drugs in vitro (24-fold increase, p<0.001). A modest increase of COX-2 mRNA was also seen after in vitro treatment of cells with cisplatin. In contrast, doxorubicin and paclitaxel caused no up-regulation in vitro, while irinotecan caused inhibition of COX-2 (2.7-fold decrease, p<0.01). These data provide a molecular rationale for clinical trials of combination chemotherapy with COX-2 inhibitors.
Notes:
Federica Di Nicolantonio, Stuart J Mercer, Louise A Knight, Francis G Gabriel, Pauline A Whitehouse, Sanjay Sharma, Augusta Fernando, Sharon Glaysher, Silvana Di Palma, Penny Johnson, Shaw S Somers, Simon Toh, Bernie Higgins, Alan Lamont, Tim Gulliford, Jeremy Hurren, Constantinos Yiangou, Ian A Cree (2005)  Cancer cell adaptation to chemotherapy.   BMC Cancer 5: 07  
Abstract: BACKGROUND: Tumor resistance to chemotherapy may be present at the beginning of treatment, develop during treatment, or become apparent on re-treatment of the patient. The mechanisms involved are usually inferred from experiments with cell lines, as studies in tumor-derived cells are difficult. Studies of human tumors show that cells adapt to chemotherapy, but it has been largely assumed that clonal selection leads to the resistance of recurrent tumors. METHODS: Cells derived from 47 tumors of breast, ovarian, esophageal, and colorectal origin and 16 paired esophageal biopsies were exposed to anticancer agents (cisplatin; 5-fluorouracil; epirubicin; doxorubicin; paclitaxel; irinotecan and topotecan) in short-term cell culture (6 days). Real-time quantitative PCR was used to measure up- or down-regulation of 16 different resistance/target genes, and when tissue was available, immunohistochemistry was used to assess the protein levels. RESULTS: In 8/16 paired esophageal biopsies, there was an increase in the expression of multi-drug resistance gene 1 (MDR1) following epirubicin + cisplatin + 5-fluorouracil (ECF) chemotherapy and this was accompanied by increased expression of the MDR-1 encoded protein, P-gp. Following exposure to doxorubicin in vitro, 13/14 breast carcinomas and 9/12 ovarian carcinomas showed >2-fold down-regulation of topoisomerase IIalpha (TOPOIIalpha). Exposure to topotecan in vitro, resulted in >4-fold down-regulation of TOPOIIalpha in 6/7 colorectal tumors and 8/10 ovarian tumors. CONCLUSION: This study suggests that up-regulation of resistance genes or down-regulation in target genes may occur rapidly in human solid tumors, within days of the start of treatment, and that similar changes are present in pre- and post-chemotherapy biopsy material. The molecular processes used by each tumor appear to be linked to the drug used, but there is also heterogeneity between individual tumors, even those with the same histological type, in the pattern and magnitude of response to the same drugs. Adaptation to chemotherapy may explain why prediction of resistance mechanisms is difficult on the basis of tumor type alone or individual markers, and suggests that more complex predictive methods are required to improve the response rates to chemotherapy.
Notes:
Mauro Moroni, Silvio Veronese, Silvia Benvenuti, Giovanna Marrapese, Andrea Sartore-Bianchi, Federica Di Nicolantonio, Marcello Gambacorta, Salvatore Siena, Alberto Bardelli (2005)  Gene copy number for epidermal growth factor receptor (EGFR) and clinical response to antiEGFR treatment in colorectal cancer: a cohort study.   Lancet Oncol 6: 5. 279-286 May  
Abstract: BACKGROUND: The antiepidermal growth factor receptor (antiEGFR) monoclonal antibodies cetuximab and panitumumab have good clinical activity in about 10% of patients with metastatic colorectal cancer that is resistant to chemotherapy. The molecular mechanisms underlying clinical response or resistance to these agents are unknown. METHODS: Tumours from 31 patients with metastatic colorectal cancer who had either an objective response (n=10) or stable disease or progressive disease (n=21) after treatment with cetuximab or panitumumab were screened for genetic changes in EGFR or its immediate intracellular effectors. Specifically, we assessed the EGFR copy number and the mutation profile of the EGFR catalytic domain and of selected exons in KRAS, BRAF, and PIK3CA. RESULTS: Eight of nine of patients with objective responses who were assessable by fluorescence in-situ hybridisation (FISH) had an increased EGFR copy number. By contrast, one of 21 non-responders assessable by FISH had an increased EGFR copy number (p<0.0001 for responders vs non-responders, Fisher's exact test). The mutation status of the EGFR catalytic domain and its immediate downstream effectors PIK3CA, KRAS, and BRAF did not correlate with disease response. In colorectal-cancer cell lines, the concentration of cetuximab that completely inhibited proliferation of cells with amplified EGFR copy number did not affect proliferation of cells with unamplified EGFR. INTERPRETATION: We propose that the response to antiEGFR treatment has a genetic basis and suggest that patients might be selected for treatment on the basis of EGFR copy number.
Notes:
2004
Federica Di Nicolantonio, Louise A Knight, Sharon Glaysher, Pauline A Whitehouse, Stuart J Mercer, Sanjay Sharma, Lisa Mills, Alison Prin, Penny Johnson, Peter A Charlton, David Norris, Ian A Cree (2004)  Ex vivo reversal of chemoresistance by tariquidar (XR9576).   Anticancer Drugs 15: 9. 861-869 Oct  
Abstract: The expression of P-glycoprotein (P-gp) has been demonstrated to confer resistance to several anticancer drugs, including anthracyclines, taxanes and vinca alkaloids. Tariquidar is a novel inhibitor of P-gp that has been shown to reverse resistance to cytotoxic drugs in tumor cell lines and mouse xenografts. We have used an ATP-based chemosensitivity assay (ATP-TCA) to compare the activity of cytotoxic drugs in combination with tariquidar against a variety of solid tumors (n = 37). The expression of P-gp was determined in a subset of solid tumor samples by immunohistochemistry (n = 16). Resistance was seen in 20 of 37 (54%) tumors tested with doxorubicin, in 27 of 34 (79%) samples tested with paclitaxel and 17 of 31 (55%) with vinorelbine. Tariquidar alone showed no activity over a wide range of concentrations up to 2 microM (n = 14). The median IC90s for doxorubicin, paclitaxel and vinorelbine, alone were 2.57, 27.4 and 15.5 microM. These decreased to 1.67 (p<0.0005), 20.6 (p<0.05) and 9.5 microM (p<0.001), respectively, in combination with tariquidar. Tariquidar also significantly decreased resistance in 14 of 20 (70%), six of 27 (22%) and six of 17 (35%) samples tested with doxorubicin, paclitaxel and vinorelbine, respectively. Immunohistochemical staining for P-gp was positive in nine of 16 (56%) samples and in all of these cases addition of tariquidar improved the activity of the cytotoxic. The results show that tariquidar is able to decrease resistance in a number of solid tumors resistant to cytotoxic drugs known to be P-gp substrates. These data support the introduction of tariquidar in combination with chemotherapy to clinical trials of patients expressing P-gp.
Notes:
Federica Di Nicolantonio, Louise A Knight, Pauline A Whitehouse, Stuart J Mercer, Sanjay Sharma, Peter A Charlton, David Norris, Ian A Cree (2004)  The ex vivo characterization of XR5944 (MLN944) against a panel of human clinical tumor samples.   Mol Cancer Ther 3: 12. 1631-1637 Dec  
Abstract: XR5944 (MLN944) is a novel DNA targeting agent with potent antitumor activity, both in vitro and in vivo, against several murine and human tumor models. We have used an ATP-tumor chemosensitivity assay to assess the ex vivo sensitivity of a variety of solid tumors (n = 90) and a CCRF-CEM leukemia cell line selected with XR5944. Differences in gene expression between the parental CCRF-CEM and the resistant subline were investigated by quantitative reverse transcription-PCR. Immunohistochemistry for topoisomerases I and IIalpha and multidrug resistance (MDR1) protein was done on those tumors for which tissue was available (n = 32). The CCRF-CEM XR5944 line showed increased mRNA levels of MDR1, major vault protein, and MDR-associated protein 1 compared with the parental line, whereas the expression of topoisomerases I, IIalpha, and IIbeta was essentially unchanged, suggesting that XR5944 is susceptible to MDR mechanisms. The median IC90 and IC50 values for XR5944 in tumor-derived cells were 68 and 26 nmol/L, respectively, 6-fold greater than in resistant cell lines. XR5944 was 40- to 300-fold more potent than the other cytotoxics tested, such as doxorubicin, topotecan, and paclitaxel. Breast and gynecologic malignancies were most sensitive to XR5944, whereas gastrointestinal tumors showed greater resistance. A positive correlation (r = 0.68; P < 0.0001) was found between the IC50 values of XR5944 and P-glycoprotein/MDR1 staining but not with either topoisomerase I or IIalpha immunohistochemistry index. These data support the rapid introduction of XR5944 to clinical trials and suggest that it may be effective against a broad spectrum of tumor types, especially ovarian and breast cancer.
Notes:
Louise A Knight, Federica Di Nicolantonio, Pauline Whitehouse, Stuart Mercer, Sanjay Sharma, Sharon Glaysher, Penny Johnson, Ian A Cree (2004)  The in vitro effect of gefitinib ('Iressa') alone and in combination with cytotoxic chemotherapy on human solid tumours.   BMC Cancer 4: Nov  
Abstract: BACKGROUND: Activation of the epidermal growth factor receptor (EGFR) triggers downstream signaling pathways that regulate many cellular processes involved in tumour survival and growth. Gefitinib ('Iressa') is an orally active tyrosine kinase inhibitor (TKI) targeted to the ATP-binding domain of EGFR (HER1; erbB1). METHODS: In this study we have used a standardised ATP-based tumour chemosensitivity assay (ATP-TCA) to measure the activity of gefitinib alone or in combination with different cytotoxic drugs (cisplatin, gemcitabine, oxaliplatin and treosulfan) against a variety of solid tumours (n = 86), including breast, colorectal, oesophageal and ovarian cancer, carcinoma of unknown primary site, cutaneous and uveal melanoma, non-small cell lung cancer (NSCLC) and sarcoma. The IC50 and IC90 were calculated for each single agent or combination. To allow comparison between samples the IndexSUM was calculated based on the percentage tumour growth inhibition (TGI) at each test drug concentration (TDC). Gefitinib was tested at concentrations ranging from 0.0625-2 microM (TDC = 0.446 microg/ml). This study represents the first use of a TKI in the assay. RESULTS: There was heterogeneity in the degree of TGI observed when tumours were tested against single agent gefitinib. 7% (6/86) of tumours exhibited considerable inhibition, but most showed a more modest response resulting in a low TGI. The median IC50 value for single agent gefitinib in all tumours tested was 3.98 microM. Interestingly, gefitinib had both positive and negative effects when used in combination with different cytotoxics. In 59% (45/76) of tumours tested, the addition of gefitinib appeared to potentiate the effect of the cytotoxic agent or combination (of these, 11% (5/45) had a >50% decrease in their IndexSUM). In 38% of tumours (29/76), the TGI was decreased when the combination of gefitinib + cytotoxic was used in comparison to the cytotoxic alone. In the remaining 3% (2/76) there was no change observed. CONCLUSION: The in vitro model suggests that gefitinib may have differential effects in response to concomitant cytotoxic chemotherapy with the agents tested during this study. The mechanism involved may relate to the effect of TKIs on growth rate versus their effect on the ability of the cell to survive the stimulus to apoptosis produced by chemotherapy.
Notes:
Federica Di Nicolantonio, Louise A Knight, Silvana Di Palma, Sanjay Sharma, Pauline A Whitehouse, Stuart J Mercer, Peter A Charlton, David Norris, Ian A Cree (2004)  Ex vivo characterization of XR11576 (MLN576) against ovarian cancer and other solid tumors.   Anticancer Drugs 15: 9. 849-860 Oct  
Abstract: XR11576 (MLN576) is a novel monophenazine with a mechanism of action that includes interaction with both topoisomerase (Topo) I and II. The aim of this study was to evaluate its cytotoxicity against fresh tumor cells taken from patients with a variety of solid tumors. Cells were obtained from 89 patients and exposed for 6 days to XR11576 alone, or in combination with doxorubicin, cisplatin, treosulfan, paclitaxel or vinorelbine. Cell survival was measured using the ATP-Tumor Chemosensitivity Assay (ATP-TCA). Immunohistochemical staining of Topo I, Topo IIalpha and MDR1 was performed on paraffin-embedded blocks in those tumors for which tissue was available (n = 49). Overall, the median IC90 and IC50 values of XR11576 in tumor-derived cells were 242 and 110 nM, respectively. In all samples XR11576 was more potent than the other cytotoxics tested. Breast and gynecological malignancies were most sensitive to XR11576, while the potency of this compound was slightly attenuated in gastrointestinal tumors, in which the median IC90 and IC50 values were 308 and 212 nM, respectively. Cases of synergism were identified when combining XR11576 with vinorelbine (nine of 30 samples) and doxorubicin (12 of 38 samples), while the addition of paclitaxel resulted in an antagonistic effect (CI50>1.2) in 38 of 42 tumors. A very modest correlation by linear regression analysis was found between the intensity of MDR1 staining and the IC50 of XR11576 (r = 0.311, p = 0.0312), but not with the IC90 (r = 0.247, NS). These data support the rapid introduction of XR11576 to clinical trials and suggest that it may be effective against a broad spectrum of tumor types.
Notes:
2003
S C Edmunds, I A Cree, F Dí Nícolantonío, J L Hungerford, J S Hurren, D P Kelsell (2003)  Absence of BRAF gene mutations in uveal melanomas in contrast to cutaneous melanomas.   Br J Cancer 88: 9. 1403-1405 May  
Abstract: The recent discovery of activating mutations in the BRAF gene in many cutaneous melanomas led us to screen the genomic sequence of BRAF exons 11 and 15 in a series of 48 intraocular (uveal) melanomas, together with control samples from three cutaneous melanomas and the SK-Mel-28 cell line, which has a BRAF mutation. The same mutation was detected in two-thirds of our cutaneous melanoma samples, but was not present in any uveal melanomas. This finding further underlines the distinction between uveal and cutaneous melanomas, and suggests that BRAF inhibitors are unlikely to benefit patients with uveal melanoma.
Notes:
Pauline A Whitehouse, Louise A Knight, Federica Di Nicolantonio, Stuart J Mercer, Sanjay Sharma, Ian A Cree (2003)  Heterogeneity of chemosensitivity of colorectal adenocarcinoma determined by a modified ex vivo ATP-tumor chemosensitivity assay (ATP-TCA).   Anticancer Drugs 14: 5. 369-375 Jun  
Abstract: Advanced colorectal cancer (CRC) has a poor prognosis with a 5-year survival of only 5% despite treatment with chemotherapeutic agents. Response rate and overall survival varies little between the commonly used single agents, although combinations achieve better outcomes. It is well established that considerable heterogeneity exists between cancers of the same tissue type, but it has been difficult to establish this for CRC. We therefore investigated the heterogeneity of chemosensitivity in CRC using a modified version of the ex vivo ATP-tumor chemosensitivity assay (ATP-TCA) capable of handling infected tumor tissue. Fifty-three specimens of primary solid or malignant effusions of CRC were tested, of which 46 (87%) were evaluable. There were considerable differences in sensitivities between individuals. The most active single cytotoxic agents in the assay were identified as 5-fluorouracil, irinotecan and mitomycin C (MMC). Cells were exposed to combinations of drugs added simultaneously at the same concentrations tested as single agents. All drug combinations achieved greater growth inhibition than drugs used alone. MMC+gemcitabine was found to be the most effective combination in 83% of specimens. The ATP-TCA has previously been shown to be a good predictor of response to chemotherapy in other tissue types. The degree of heterogeneity demonstrated from these results suggests that the ATP-TCA could be used to identify patients who might benefit from specific chemotherapeutic agents alone or in combination.
Notes:
Sanjay Sharma, Michael H Neale, Federica Di Nicolantonio, Louise A Knight, Pauline A Whitehouse, Stuart J Mercer, Bernard R Higgins, Alan Lamont, Richard Osborne, Andrew C Hindley, Christian M Kurbacher, Ian A Cree (2003)  Outcome of ATP-based tumor chemosensitivity assay directed chemotherapy in heavily pre-treated recurrent ovarian carcinoma.   BMC Cancer 3: Jul  
Abstract: BACKGROUND: We wished to evaluate the clinical response following ATP-Tumor Chemosensitivity Assay (ATP-TCA) directed salvage chemotherapy in a series of UK patients with advanced ovarian cancer. The results are compared with that of a similar assay used in a different country in terms of evaluability and clinical endpoints. METHODS: From November 1998 to November 2001, 46 patients with pre-treated, advanced ovarian cancer were given a total of 56 courses of chemotherapy based on in-vitro ATP-TCA responses obtained from fresh tumor samples or ascites. Forty-four patients were evaluable for results. Of these, 18 patients had clinically platinum resistant disease (relapse < 6 months after first course of chemotherapy). There was evidence of cisplatin resistance in 31 patients from their first ATP-TCA. Response to treatment was assessed by radiology, clinical assessment and tumor marker level (CA 125). RESULTS: The overall response rate was 59% (33/56) per course of chemotherapy, including 12 complete responses, 21 partial responses, 6 with stable disease, and 15 with progressive disease. Two patients were not evaluable for response having received just one cycle of chemotherapy: if these were excluded the response rate is 61%. Fifteen patients are still alive. Median progression free survival (PFS) was 6.6 months per course of chemotherapy; median overall survival (OAS) for each patient following the start of TCA-directed therapy was 10.4 months (95% confidence interval 7.9-12.8 months). CONCLUSION: The results show similar response rates to previous studies using ATP-TCA directed therapy in recurrent ovarian cancer. The assay shows high evaluability and this study adds weight to the reproducibility of results from different centres.
Notes:
Stuart J Mercer, Shaw S Somers, Louise A Knight, Pauline A Whitehouse, Sanjay Sharma, Federica Di Nicolantonio, Sharon Glaysher, Simon Toh, Ian A Cree (2003)  Heterogeneity of chemosensitivity of esophageal and gastric carcinoma.   Anticancer Drugs 14: 6. 397-403 Jul  
Abstract: Esophageal and gastric cancer have a poor prognosis, and chemotherapy is rarely of long-term benefit. This may be related in part to heterogeneity of chemosensitivity and to constitutive resistance to individual cytotoxic drugs. This study aimed to demonstrate the degree of heterogeneity of chemosensitivity between tumors. We have examined the heterogeneity of chemosensitivity in esophageal and gastric cancer specimens (n=85) using an ex vivo ATP-based chemosensitivity assay (ATP-TCA). A variety of chemotherapeutic agents were tested. Sixty-four specimens were endoscopic biopsy samples; the remainder were from resection specimens. Cells were obtained from 62 specimens (73%). Eight assays were infected due to contamination/infection of the biopsy material, giving an evaluability rate of 87%. Analysis of the data showed considerable heterogeneity of chemosensitivity. The most active single agents identified by the assay were mitomycin C (56% sensitivity) and 5-fluorouracil (5-FU; 42% sensitivity). Exposure of tumor cells to combinations of drugs showed ECF (epirubicin, cisplatin, 5-FU) and mitomycin C+5-FU to be moderately active regimens. Other experimental drug combinations showed greater activity. There is a marked heterogeneity of chemosensitivity in esophageal and gastric cancers. The degree of heterogeneity observed suggests that the ATP-TCA could be used to individualize chemotherapy by selecting agents for particular patients. This approach provides the rationale for a trial of ATP-TCA-directed therapy to determine whether individualization of chemotherapy might improve patient response and survival.
Notes:
P A Whitehouse, S J Mercer, L A Knight, F Di Nicolantonio, A O'Callaghan, I A Cree (2003)  Combination chemotherapy in advanced gastrointestinal cancers: ex vivo sensitivity to gemcitabine and mitomycin C.   Br J Cancer 89: 12. 2299-2304 Dec  
Abstract: Advanced or metastatic disease is common in both oesophagogastric and colorectal cancers, with poor 5-year survival despite palliative chemotherapy. We have investigated the sensitivity of gastrointestinal tumours to gemcitabine in combination with mitomycin C (GeM), using a modified ex vivo ATP-based tumour chemosensitivity assay (ATP-TCA). Tumour material from 41 colorectal and 22 oesophagogastric cancers were assessed. The GeM combination showed variable but definite activity in most of the samples tested. The results show that GeM achieves >95% inhibition at concentrations within the range achievable clinically in 60% of colorectal tumours (21 out of 35) and 38% of oesophagogastric tumours (five out of 13) tested. We did not identify any significant difference in sensitivity using concurrent or sequential exposure of tumour-derived cells to these two drugs. The results from this study suggest that GeM may be a useful combination in the treatment of advanced gastrointestinal malignancy.
Notes:
Thomas Schöndorf, Rainer Neumann, Carolin Benz, Martina Becker, Marion Riffelmann, Uwe-Jochen Göhring, Judith Sartorius, Carl-Heinz Wirsing von König, Martina Breidenbach, Markus M Valter, Markus Hoopmann, Federica Di Nicolantonio, Christian M Kurbacher (2003)  Cisplatin, doxorubicin and paclitaxel induce mdr1 gene transcription in ovarian cancer cell lines.   Recent Results Cancer Res 161: 111-116  
Abstract: The clinical observation of the multidrug resistance (MDR) phenotype is often associated with overexpression of the mdrl gene, in particular with respect to ovarian cancer. However, until now the mdrl-inducing potential of commonly used antineoplastics has been only incompletely explored. We performed short-term cultures of six ovarian cancer cell lines (MZOV4, EF027, SKOV3, OAW42, OTN14, MZOV20) exposed to either blank medium or cisplatin, doxorubicin or paclitaxel at concentrations related to the clinically achievable plasma peak concentration. A highly specific quantitative real-time RT-PCR was used to detect the Mdr1 transcripts. Mdrl mRNA contents were calibrated in relation to coamplified GAPDH mRNA. Mdrl mRNA was detectable in each cell line. In 13 out of 18 assays (72%) the specific anticancer drug being tested induced mdr1 transcription. No decrease in mdr1 mRNA concentration was observed. Our data suggest that mdr1 induction by antineoplastics is one of the reasons for failure of ovarian cancer therapy but may vary individually.
Notes:
Federica Di Nicolantonio, Michael Neale, Zerrin Onadim, John L Hungerford, Judith L Kingston, Ian A Cree (2003)  The chemosensitivity profile of retinoblastoma.   Recent Results Cancer Res 161: 73-80  
Abstract: Retinoblastoma is a rare malignant tumour of the developing retina with an incidence of 1 in 20,000 live births in all human races. Chemotherapy is used in retinoblastoma as adjuvant therapy to prevent the growth of metastases and to treat metastatic disease once this has become clinically apparent. Current regimens are based on empirical drug combinations, and few clinical trials have been conducted because of the rarity of this tumour. Chemosensitivity testing offers a way of testing a large number of agents against tumours. The ATP-based chemosensitivity assay (ATP-TCA) has already helped to design new regimens for melanoma and breast and ovarian cancer. Primary retinoblastoma tumour material was obtained from 10 eyes, 7 of which contained sufficient viable cells for ATP-TCA. The results show very high sensitivity to single agents, particularly cisplatin, doxorubicin and vinca aLkaloids. Of the anti-metabolites tested, 5-FU is relatively disappointing (although still active), and gemcitabine shows considerable activity consistent with a cytotoxic effect. The shape of the inhibition curves is interesting. There is a plateau effect with the topoisomerase inhibitors and vinblastine, which is not present with the cisplatin. One tumour was much more resistant than the others tested, particularly to vinblastine but also to the topoisomerase inhibitors, which failed to achieve complete kill at any concentration tested, consistent with a multidrug resistance phenotype. Of the combinations (VAC and VEC), the VAC regimen looks marginally more active in the more resistant of the two cases tested to date. These data confirm that retinoblastoma is a rapidly growing malignancy that is very susceptible to cytotoxic drugs of all types. Chemosensitivity testing provides a practical method of testing new regimens before clinical trials in retinoblastoma patients.
Notes:
2002
Ian A Cree, Louise Knight, Federica Di Nicolantonio, Sanjay Sharma, Tim Gulliford (2002)  Chemosensitization of solid tumors by modulation of resistance mechanisms.   Curr Opin Investig Drugs 3: 4. 634-640 Apr  
Abstract: The number of drugs available for chemotherapy is growing exponentially, and this trend looks set to continue. Chemosensitization strategies use the administration of one drug or agent to render cancer cells more susceptible to a second agent. Modulation of resistance mechanisms due to xenobiotic membrane pumps such as the multidrug resistant proteins, MDR1/P-glycoprotein or MRP is feasible and a number of new agents have been produced to inhibit drug efflux resulting from expression of these molecules. However, tumor cells may express or upregulate more than one such molecule at one time, and this approach is unlikely to benefit every patient. Detoxification mechanisms mediated by glutathione conjugation or metallothionein are also responsible for resistance--the former has been linked to MRP-mediated resistance. Again, modulation is possible but may increase the toxicity of drugs to normal tissues and an increased therapeutic index is not guaranteed. Tumors exposed to DNA damaging agents often upregulate DNA repair mechanisms and this contributes to resistance. Different pathways perform the repair of different forms of DNA damage, and it is difficult to inhibit all of these. Nevertheless, inhibition of DNA repair can re-sensitize tumors to chemotherapy and is increasingly exploited. One of the most successful and widely used approaches is to combine gemcitabine with an alkylating or platinating agent. While gemcitabine may inhibit DNA polymerases directly, this cytidine analog is also likely to be incorporated by DNA repair leading to activity against non-cycling cells, which form the majority of the neoplastic cell population in most solid tumors. Oncologists should take account of potential resistance mechanisms when treating patients: it is often feasible to design combinations with old or new drugs which exploit these apparent weaknesses to the patient's advantage.
Notes:
Ian A Cree, Louise Knight, Federica Di Nicolantonio, Sanjay Sharma, Tim Gulliford (2002)  Chemosensitization of solid tumor cells by alteration of their susceptibility to apoptosis.   Curr Opin Investig Drugs 3: 4. 641-647 Apr  
Abstract: Chemosensitization strategies use the administration of one drug or agent to render cancer cells more susceptible to a second agent. Usually this involves enhanced drug metabolism, improvement of drug uptake or blockage of resistance mechanisms. Alteration of the susceptibility of cancer cells to apoptosis, the process of individual cell death by which many chemotherapeutic drugs act, shows particular promise for therapy in the future, and is the focus of this review. The dependence of cancer cells on non-neoplastic cells to form solid tumors allows anti-angiogenic therapy to be used in conjunction with chemotherapy to increase the therapeutic index. Chemosensitization strategies are set to become increasingly important in cancer therapy, allowing rational design of synergistic drug combinations at an earlier stage in drug development.
Notes:
Federica Di Nicolantonio, Michael H Neale, Louise A Knight, Alan Lamont, Geraldine E Skailes, Richard J Osborne, Rosanne Allerton, Christian M Kurbacher, Ian A Cree (2002)  Use of an ATP-based chemosensitivity assay to design new combinations of high-concentration doxorubicin with other drugs for recurrent ovarian cancer.   Anticancer Drugs 13: 6. 625-630 Jul  
Abstract: Liposomal doxorubicin (Caelyx/Doxil) has been shown to be active in around 20% of recurrent ovarian cancers. As yet, there is little clinical data on combinations of existing agents with liposomal doxorubicin, despite considerable clinical experience with soluble doxorubicin in combination. In this study, we have used an ATP-based tumor chemosensitivity assay to determine the relative efficacy of high concentrations of doxorubicin tested in combination with cisplatin, treosulfan, 5-fluorouracil (5-FU) or vinorelbine against cells obtained from recurrent ovarian tumor tissue. The results show little enhancement of the efficacy of high concentrations of doxorubicin by 5-FU, cisplatin, or treosulfan. However, vinorelbine+liposomal doxorubicin showed additive inhibition, and this combination is worthy of further testing in clinical trials.
Notes:
2001
L Cantoni, F Di Nicolantonio, D Barelli, M Rizzardini, F De Matteis (2001)  Modulation of cerebellar and hepatic nitric oxide synthase by exogenous arginine and endotoxin.   Nitric Oxide 5: 2. 198-207 Apr  
Abstract: We have studied in mice the effect of treatment with exogenous arginine and/or LPS by monitoring serum nitrite/nitrate levels and by investigating the response of cerebellar and liver nitric oxide synthase (NOS). We measured NOS activity in cerebellar extracts while changes in iNOS mRNA were followed in the liver since direct assay of NOS activity proved unreliable with this tissue. In fact, liver and cerebellum extracts were both very active in converting arginine into a citrulline-like metabolite, but only cerebellum conversion was dependent on addition of NADPH and inhibitable by N(G)-methyl-l-arginine. Treatment with LPS, on its own, increased serum nitrite/nitrate levels at 5 and 20 h after injection, while treatment with LPS and arginine produced nitrite/nitrate levels in the serum even greater at 5 h, but significantly lower at 20 h. Liver iNOS mRNA levels were markedly increased by LPS, and this effect was significantly decreased when mice were also given exogenous arginine. A stimulatory effect of LPS was also found on NOS activity in the cerebellum, where a very small stimulation may have also been caused by arginine feeding. These findings indicate that LPS stimulates NOS expression/activity both in the cerebellum and in the liver and suggest a complex pattern of modulation of iNOS by arginine, with NO being first produced in excess and then downregulating iNOS expression.
Notes:

Conference papers

2013
Federica Di Nicolantonio (2013)  How to Assess Potential Biomarkers in Clinical Trials   In: ASCO 2013 Gastrointestinal Cancers Symposium 62-3  
Abstract: Cancer onset and progression is caused by the accumulation of genomic changes, which include mutations, amplifications and deletions, genome rearrangements, as well as epigenetic modulation of gene expression. This surge of cancer genomics data has allowed the identification of molecular subtypes of cancer, and has guided the design of novel inhibitors to specifically target those genes and pathways driving the malignant phenotype. It is therefore logical that molecular biomarkers should be used to drive the successful development of targeted drugs as well as to tailor therapy to the individual patient. Indeed, recent initiatives from the US Food and Drug Administration (FDA) have acknowledged the role of well-characterized biomarkers during drug development. I will focus on strategies to assess those biomarkers that are most relevant for identifying patients who are likely to respond to a given therapy. An overview of molecular markers that can be used for measuring the efficacy and detecting the onset of resistance to a given therapy will also be provided. These two measures are key for selecting the right drug for the right patient, regardless of whether they are applied during drug development or in the post-approval phase. Prospective or retrospective assessment of biomarkers? Biomarkers can be assessed in a prospective or retrospective manner – or even using a combination of both approaches. Prospective biomarkers are recommended when very robust preclinical data are available and their prevalence is expected to occur in a very small subpopulation (<5%). As an example, the prevalence of HER2 amplification in mCRC is around 2-3% 1 and the combination of an antibody (trastuzumab) and small molecule receptor tyrosine kinase inhibitor (lapatinib) was able to induce tumor regression in HER-2 amplified patient-derived tumor xenografts. 2 This has provided the rationale for a phase II trial testing the activity of trastuzumab plus lapatinib in chemorefractory metastatic colorectal cancer (mCRC) patients preselected on tumor HER2 amplification (HERACLES; HER2 Amplification for Colo-rectaL cancer Enhanced Stratification; EUCTR2012-002128-33-IT). The rationale for this study is based on the assumption that in a small subset of CRC tumors HER2 amplification is the driver genetic lesion. Despite the clear advantage of conducting a small study in a highly selected population, this approach has some limitations, as it does not offer the opportunity to look for efficacy outside the ‘target’ population or to refine the definition of ‘target’ population. Additionally, it requires the existence of a robust and reproducible biomarker screening assay for HER2 gene amplification in CRC samples. In the absence of a strong scientific rationale, patient enrichment is not recommended and ‘all-comers’ should be recruited. While such studies are conducted in an unselected population, biomarkers can still be assessed retrospectively. Retrofitting biomarkers to clinical programs has seldom been successful. As rare example, the identification of KRAS mutations as a negative predictor of response to anti-EGFR therapies in mCRC patients became mainly apparent from retrospective analyses. Although the strategy was ultimately successful, costly phase II/III studies have exposed hundreds of patients to a drug from which they did not benefit. In other instances, combination of retrospective and prospective approaches can be considered. In early drug development, dose-escalation studies can be performed in all-comers, while the use of a molecular test can be restricted to an expansion cohort. In more advanced phases, an initial equal randomization can be followed by an adaptive randomization. Finally, in the post-genomics era, multi-marker platform studies, such as the phase II (potentially phase III) FOCUS 4 trial are particularly suited to stratify patients on several molecular markers and rationally test selected targeted drugs for single-agent or combined-agent activity. FOCUS 4 has an adaptive design, which allows for efficient incorporation of new information or drugs into the trial. Patients will be stratified and treated with targeted agents on the basis of developing – but not yet validated – biomarker data, which currently include mutations in oncogenes such as KRAS, BRAF, NRAS and PIK3CA; expression/ sequencing/ methylation of the PTEN oncosuppressor gene; expression (mRNA) levels of known modulators of EGFR-MEK signaling (EREG, DUSP4, DUSP6); and immunohistochemical staining for DNA mismatch repair proteins (MLH1, MSH2, MSH6, PMS2). Practical implementation of biomarker testing Insight of CRC pathogenesis and practical issues should be taken in careful consideration while planning the incorporation of biomarkers in clinical trials. CRCs are ‘smart tumors’ carrying several genomic aberrations in each individual sample.1 Co-existence of multiple changes likely result in redundancy of oncogenic signaling pathways, thereby limiting the usefulness of single-gene based biomarkers in predicting response to target therapies. Tumor genomic alterations may evolve over time and under treatment with chemotherapy. While a number of studies have demonstrated good concordance between primary and metastatic lesions in the mutational status of selected key oncogenes, such as KRAS, BRAF or PIK3CA, 3,4 full exome sequencing has revealed genetic differences in primary CRCs and their subsequent hepatic metastases, 5 questioning the relevance of archival primary resection specimens to the metastatic setting. To address this issue, collection of cancer biopsies in clinical trials has become increasingly mandatory to allow for prospective biomarkers testing on samples which best reflect the patient current situation at the time of enrolment. However, direct and repeated tumor biopsy may be difficult or otherwise unattainable. Additionally – and perhaps more importantly– sampling of a single lesion could not be representative of the cancer genomic landscape, since it has recently been shown that not all mutations are ubiquitously present in every region of a tumor. 6 To overcome these limitations, we and others have recently shown that cell free circulating tumor DNA (cfDNA) can be isolated from mCRC patients’ plasma samples and used to study KRAS genetic variants before and during treatment with anti-EGFR targeted therapies. 7-9 Peripheral blood drawing requires a minimally invasive procedure and cfDNA contains genetic information which is representative of all the tumor subclones present in the body (without needing access to the actual samples). Next-generation sequencing technologies could ultimately be employed to perform whole exome/genome analysis of cfDNA (and/or circulating tumor cells), thus capturing the genetic complexity of all cancer cells within an individual patient. The ability to conduct these analyses in a reliable, reproducible, fast and cost-effective manner should allow for their incorporation in clinical protocols, enabling us to assess biomarkers at baseline as well as over the course of therapy. Summary Despite the intensive research efforts invested in drug and biomarkers development, only very few of them have entered clinical practice. Innovative clinical trial designs incorporating biomarkers from early phases are expected to reduce drug attrition rates. As we rapidly move from single gene-based biomarker to multi-parameter and eventually to genome testing, we would likely face new biological and practical challenges. These will include patient intratumor heterogeneity; access to longitudinal serial samples; clinical implementation of robust and cost-effective technologies for cancer genomics; as well as the ability to perform integrated genomic data analysis in a timely fashion. References 1. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487:330-7, 2012 2. Bertotti A, Migliardi G, Galimi F, et al: A molecularly annotated platform of patient-derived xenografts ("xenopatients") identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discovery 1:508-23, 2011 3. Artale S, Sartore-Bianchi A, Veronese SM, et al: Mutations of KRAS and BRAF in primary and matched metastatic sites of colorectal cancer. J Clin Oncol 26:4217-9, 2008 4. Vakiani E, Janakiraman M, Shen R, et al: Comparative genomic analysis of primary versus metastatic colorectal carcinomas. J Clin Oncol 30:2956-62, 2012 5. Vermaat JS, Nijman IJ, Koudijs MJ, et al: Primary colorectal cancers and their subsequent hepatic metastases are genetically different: implications for selection of patients for targeted treatment. Clin Cancer Res 18:688-99, 2012 6. Gerlinger M, Rowan AJ, Horswell S, et al: Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 366:883-92, 2012 7. Diaz LA, Jr., Williams RT, Wu J, et al: The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 486:537-40, 2012 8. Misale S, Yaeger R, Hobor S, et al: Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature advance online publication, 2012 9. Spindler KL, Pallisgaard N, Vogelius I, et al: Quantitative cell-free DNA, KRAS, and BRAF mutations in plasma from patients with metastatic colorectal cancer during treatment with cetuximab and irinotecan. Clin Cancer Res 18:1177-85, 2012
Notes:
 BRAF, PIK3CA and KRAS mutations and loss of PTEN expression impair response to EGFR-targeted therapies in metastatic colorectal cancer   In: 100th Annual Meeting of the American Association for Cancer Research AACR  
Abstract: INTRODUCTION. EGFR-mediated signaling involves two main intracellular cascades: on one side KRAS activates BRAF, which in turn triggers the mitogen-activated protein kinases. On the other, membrane localization of the lipid kinase PIK3CA counteracts PTEN and promotes AKT1 phosphorylation, thereby activating a parallel intracellular axis. Constitutive activation of KRAS bypasses the corresponding signaling cascade and, accordingly, patients with metastatic colorectal cancer (mCRC) bearing KRAS mutations are clinically resistant to EGFR-targeted monoclonal antibodies (moAbs) cetuximab or panitumumab. However, mutations in KRAS can only explain 30-40% resistant cases. Recent reports indicate that alterations in other members of the EGFR signaling cascade, such as BRAF mutations and deregulation of the PIK3CA/PTEN pathway, could also drive resistance to anti-EGFR therapy. METHODS. To clarify the relative contribution of these molecular alterations to response to anti EGFR MoAbs, we performed a comprehensive analysis of KRAS, BRAF, PIK3CA gene mutations and loss of PTEN expression in tumor samples from a cohort of 132 mCRC patients treated with cetuximab or panitumumab containing regimens. RESULTS. KRAS, BRAF and PIK3CA mutations were present in 35, 11 and 17 cases, respectively, while loss of PTEN was found in 41 cases. KRAS and BRAF mutations were mutually exclusive; KRAS and PIK3CA alterations were concomitantly present in 2 samples, while BRAF and PIK3CA mutations co-occurred in 7 cases. In univariate analysis, KRAS mutations were associated with resistance to cetuximab or panitumumab (P=0.026), although 2 patients carrying a KRAS G13D mutated tumor achieved objective response. The presence of BRAF or PIK3CA mutations correlated with lack of clinical response: none of the responders harbored genetic alterations in either of these genes, and only 1/41 patients with PTEN deficient tumors displayed a partial response (P < 0.01). BRAF and/or PIK3CA mutated patients displayed also significantly shorter progression-free survival (P < 0.005). BRAF mutations were a prognostic factor for decreased overall survival (P < 0.01). In multivariate analysis, mutations of KRAS and loss of PTEN were the only independent predictors of lack of objective response (P=0.0014 and 0.0001, respectively). Cox multivariate analysis for survival demonstrated that mutations of BRAF and loss of PTEN are independently associated with decreased overall survival (P=0.011 and 0.012, respectively), with a trend toward statistical significance for KRAS mutations (P=0.054). The presence of mutations in at least one gene among KRAS, BRAF or PIK3CA was significantly associated with lack of response to EGFR targeted moAbs (P<0.0001; specificity =0.96; range 0.87-0.99), and negatively impacted on both progression-free and overall survival. When the expression of PTEN and mutations of KRAS, BRAF or PIK3CA were concomitantly ascertained, over 70% of mCRC patients unlikely to respond to EGFR moAbs could be identified. CONCLUSIONS. Comprehensive molecular dissection of the EGFR signalling pathways should be considered to select mCRC patients eligible for anti-EGFR moAbs therapies. Prospective studies are therefore needed to address the potential role of BRAF, PIK3CA and PTEN to help selection and stratification of mCRC patients treated with anti-EGFR therapies.
Notes:
Powered by PublicationsList.org.